Skip to main content
Mediators of Inflammation logoLink to Mediators of Inflammation
. 2012 Nov 7;2012:546786. doi: 10.1155/2012/546786

Mechanism of Inflammation in Age-Related Macular Degeneration

Francesco Parmeggiani 1,*, Mario R Romano 2,3, Ciro Costagliola 2, Francesco Semeraro 4, Carlo Incorvaia 1, Sergio D'Angelo 1, Paolo Perri 1, Paolo De Palma 1, Katia De Nadai 1,5, Adolfo Sebastiani 1
PMCID: PMC3504473  PMID: 23209345

Abstract

Age-related macular degeneration (AMD) is a multifactorial disease that represents the most common cause of irreversible visual impairment among people over the age of 50 in Europe, the United States, and Australia, accounting for up to 50% of all cases of central blindness. Risk factors of AMD are heterogeneous, mainly including increasing age and different genetic predispositions, together with several environmental/epigenetic factors, that is, cigarette smoking, dietary habits, and phototoxic exposure. In the aging retina, free radicals and oxidized lipoproteins are considered to be major causes of tissue stress resulting in local triggers for parainflammation, a chronic status which contributes to initiation and/or progression of many human neurodegenerative diseases such as AMD. Experimental and clinical evidences strongly indicate the pathogenetic role of immunologic processes in AMD occurrence, consisting of production of inflammatory related molecules, recruitment of macrophages, complement activation, microglial activation and accumulation within those structures that compose an essential area of the retina known as macula lutea. This paper reviews some attractive aspects of the literature about the mechanisms of inflammation in AMD, especially focusing on those findings or arguments more directly translatable to improve the clinical management of patients with AMD and to prevent the severe vision loss caused by this disease.

1. Introduction

Age-related macular degeneration (AMD) is a common disease of the central area in the ocular posterior segment, known as the macula lutea. This retinal area is essential for the vision of fine details and the image resolution, capturing the greatest focus of the external light stimuli. In the human macula, there are several recognizable main structures: the neuroretina (NR), composed by the inner neurosensory layer and outer photoreceptor cell layer with the underlying retinal pigment epithelium (RPE); this latter structure is separated from the choriocapillaris (CC) by the Bruch's membrane (BM), a modified basement stratum. The retina contains two types of photoreceptors, rods and cones. The rods are more numerous (about 120 million) and are more sensitive than the cones, being responsible for vision at low light levels (scotopic vision). They are not sensitive to colors and characterized by low spatial acuity. Conversely, the 6 to 7 million cones are active at higher light levels (photopic vision), are capable of color vision, and are responsible for high spatial acuity. The center of the macula, called fovea centralis, is an avascular zone exclusively populated by cones (Figures 1(a) and 1(b) [4]). AMD is due to multifaceted degenerative disorders involving the NR-RPE-BM-CC complex at the level of macular region [13].

Figure 1.

Figure 1

Normal human macula. (a) Schematic cross-sectional illustration of the macular outer segment and, in particular, the cells of the retinal pigment epithelium ((1) nucleus, (2) mitochondria, (3) ribosomes, (4) lysosomes, (5) Golgi apparatus, (6) melanosomes, (7) lipofuscin granules, (8) zonula occludens, (9) photoreceptor (cone), (10) outer segment of cones, (11) phagocytosis of photoreceptorial discs, (12) phagosome, (13) Bruch's membrane, and (14) choriocapillaris). (b) Fluorescein angiography of the macula with its foveal avascular zone (extracted and modified from [4]).

AMD represents the main cause of legal blindness or low vision in those developed countries with the longest life expectance, especially affecting the elderly people of European descent [3]. In North America, Europe, and Australia, AMD accounts for up to 50% of all cases of central blindness [5], approximately reaching a prevalence of 3% among the general adult population [6]. In the United States, about 15% of people older than 80 years of age were estimated to have AMD on 2000, and this number is expected to rise in the next years reaching more than 2.95 million people with AMD in 2020 [7].

Numerous and heterogeneous pathological processes are likely to predispose an individual to AMD, which is considered an extremely complex, multifactorial disease. Aging represents its primary determinant, while environmental factors such as cigarette smoking [8, 9], dietary habits [1012], and phototoxic exposure [1315] contribute to significantly increase the risk of AMD occurrence, together with several gene polymorphisms [1622]. In a population-based twin study including both concordant/discordant and monozygotic/dizygotic sibling pairs, Seddon and coworkers have evaluated the relative contribution of heredity and environment to AMD etiology, concluding that heritability estimates for AMD are remarkable and range from 46% to 71% [23]. More recently, the same research group has indicated that individual genotypic susceptibility interacts with behavioral and nutritional factors in the etiology of AMD by means of various epigenetic mechanisms [24], further supporting the importance of the epigenetics into AMD investigations [25, 26]. These data, along with findings of genome-wide association studies, emphasize the presence of an important rationale to practice the search for AMD-related gene variants [27, 28], despite the unavoidable efforts required to plan genetic analyses of a complex disease with late onset. In particular, remarkable correlations were documented between common or rare immunological/inflammatory gene polymorphisms and AMD, unequivocally indicating the involvement of inflammation and immune-mediated processes (complement activation) in the pathogenesis of this disease [21, 2932]. Thus, although AMD is not considered a classic inflammatory disease, immunocompetent cells, such as macrophages and lymphocytes, are present in the chorioretinal tissues affected by AMD [33, 34]. Moreover, peculiar signs of abnormalities/dysregulation of innate immune system are observed in eyes with AMD principally at the level of the complement pathway, including complement components C3a and C5a, C5 and C5b-9 terminal complement complex, complement regulators or inhibitors, that is, complement factor H (CFH), vitronectin and clusterin, complement receptor 1 (CR1, also called CD35), membrane cofactor protein (MCP, also called CD46), and decay accelerating factor (DAF, also called CD55), but also at the level of C-reactive protein (CRP) [18, 3541]. In particular, activation products C3a, C5a, and C5b-9 are also systemically elevated in patients suffering from AMD [4245]. In the course of AMD, several immunopathological phenomena occur within the NR-RPE-BM-CC complex of the macular area, especially due to the pathophysiologic effects of complement system, which have a main role in the parainflammation of the aging retina [4652]. Herein, we briefly review the literature on the involvements of inflammation in AMD, highlight the possible environmental, genetic, and/or epigenetic interactions, and discuss those therapeutic approaches potentially able to modulate inflammatory pathways and more directly translatable to the management of AMD patients.

2. Parainflammation and Age-Related Macular Degeneration

Parainflammation is defined as a condition of tissue adaptive response to noxious stress or malfunction, and it has features which are considered as intermediate between normal/basal and inflammatory/acute states. Although the physiological purposes of normal parainflammation are to preserve tissues homeostasis and to restore their functionality, when a tissue is exposed to stress and/or malfunction for a prolonged period, it is implicated in both initiation and progression of many human age-related disorders, such as AMD [47, 53]. The risks of degenerative diseases, at least partially related to the pathophysiologic para-inflammatory response, are especially relevant in those tissues functionally dependent on nonproliferative cells and characterized by very high metabolism and other oxidative stress, such as the macular retina. In humans, the retina is a highly differentiated neuroectodermal tissue, in which an outer layer of photoreceptors, two layers of neuronal cells bodies, and two layers of synapses are present. The NR, together with RPE cells, forms the intraocular functional unit of the visual system. Like the components of central nervous system (brain and spinal cord) and several other tissues, retina also undergoes many pathophysiologic modifications with age. Because of cell and tissue damage/malfunction, mainly due to accumulative oxidative and metabolic changes in NR-RPE-BM-CC complex induced by reactive oxygen species (ROS), the vision sensitivity progressively declines during the aging process. In accordance with the “free radical theory of aging,” originally expressed by Harman in 1956, age-related degeneration is basically caused by an imbalance between ROS-induced tissue damages and repair/remodelling processes [54]. This concept seems to be extremely important for human AMD; in fact, its main risk factors include increased age, smoking, augmented body mass index, phototoxicity and inflammation [815, 55], and all these factors augment ROS generation [14, 5662]. Moreover, exactly the innate immune system, which plays a key role in tissue repair/remodeling processes, is also the same one that is more interconnected with AMD susceptibility and etiopathogenesis starting, respectively, from genotypic [21, 2932] and phenotypic [18, 3545] points of view. Particularly, the outer photoreceptor/RPE/MB complex, that is, the site of onset of the elementary AMD lesions (drusen), is considered more prone to oxidative stress because of both its proximity with the highly variable choroidal hemodynamics and its continuous exposition to photooxidation due to light stimuli [1315, 63, 64]. In fact, unregulated blood flow may increase the fluctuations of tissue oxygen concentration, leading to elevated ROS generation by the mitochondria [63, 64]. Likewise, photooxidation in photoreceptors is associated with complement activation [65], which can increase membrane attack complex formation, an important trigger of those apoptotic processes inducing nonlethal, retinal degeneration [6668]. ROS augmentation can also trigger angiogenic signaling that has a crucial role in the occurrence of the more severe complication of AMD, that is, choroidal neovascularization (CNV) [6972]. In other words, several factors, linked to AMD etiopathogenesis, lead to increased ROS generation and can mediate apoptosis and angiogenesis, which are more implicated in the atrophic and neovascular AMD forms, respectively [14, 46, 6372]. Finally, the critical position of complement must be, once again, emphasized. In fact, dysregulation of complement pathways can lead to that autologous damage which, at the macular level, is manifested by the development of drusen. Starting from this rational (even if notional) point of view, the earliest hallmarks of AMD may act as foci of chronic inflammation [49, 52, 73].

During the normal aging, in the NR, the number of neuronal and ganglion cells decreases, as also happens in the case of RPE cells which generally suffer the greatest losses in the macular and surrounding areas. Lipofuscin, the main aging-associated retinal end product, accumulates in the RPE cells with age, and its autofluorescent properties are routinely used in the clinical practice for the diagnostic imaging of various macular disorders. Another crucial age-related retinal change is the BM alteration, characterized by an increased thickness, accumulations of basal laminar deposits and/or drusen formation, and frequently accompanied by pigmentary irregularities due to RPE cell hypertrophy, hyperplasia, or atrophy. Usually, all these occurrences are more evident at the posterior pole in comparison with retinal periphery. In addition, both biochemical constitutions and biophysical properties of the BM modify with age, also influencing a further RPE cell dysfunction as well as noticeable CC disorders. Although the retina has been traditionally considered as an “immunologically privileged” tissue, at present it is known to have an endogenous immune system, actively coordinated by immunocompetent cells (microglia and dendritic cells), along with a rare population of perivascular macrophages; moreover, also RPE cells possess a variety of immunological functions. Retinal microglia and RPE cells, together with choroidal macrophages/dendritic cells, physiologically play an essential role in retinal homeostasis [47, 7478]. In the aging retina, all these elements represent important factors in both dealing with the retinal malfunctions and restoring retinal homeostasis or rebalancing the homeostatic points. Several sight-threatening retinal diseases have a higher prevalence among the elderly persons, but the most common of these is AMD that can be diagnosed in its early (Figures 2(a) and 2(b) [4]) or intermediate (Figures 3(a) and 3(b) [4]) drusen/RPE-atrophy/pigmentary forms, as well as in its advanced forms, that is, geographic atrophy and neovascular AMD (Figures 4(a)4(d)) [3]. Even if the clinical pattern of the above-mentioned types of AMD significantly differs, low-grade/subclinical degree of inflammation (parainflammation) is implicated in every AMD forms, reaching a high level when maculopathy is complicated by CNV development [71]. In neuroretinal structure, para-inflammatory modifications are characterized by the breakdown of blood-retinal barrier, microglial activation, and subretinal migration, whereas, in the choroid they become evident with an increased number of CD45+ CRIg+ macrophages, morphologic abnormalities of melanocytes, tissue's thickening, and fibrosis. At the retinal/choroidal interface, these AMD-related changes are particularly manifested by complement activation in RPE-BM cells and microglia accumulation in subretinal space [47, 48, 71]. Insightful knowledge on the mechanisms of retinal parainflammation, as well as of complement dysregulation, is fundamental to comprehensively understand the pathogenesis of AMD and to develop better curative therapeutic strategies for the different forms of this harmful disease.

Figure 2.

Figure 2

Early age-related macular degeneration. (a) Schematic cross-sectional illustration of the macula with an early stage of the disease ((1) drusen). (b) Fluorescein angiography of the macula affected by an early form of the disease (nonconfluent hard drusen); in this eye, the best best-correct visual acuity was 20/20 (Snellen equivalent) (extracted and modified from [4]).

Figure 3.

Figure 3

Intermediate age-related macular degeneration. (a) Schematic cross-sectional illustration of the macula with an intermediate stage of the disease ((1) drusen; (2) atrophy of a cell of the retinal pigment epithelium; (3) hypertrophy or hyperplasia of a cell of the retinal pigment epithelium; (4) a normal cell of the retinal pigment epithelium). (b) Fluorescein angiography of the macula affected by an intermediate form of the disease (confluent soft drusen and pigmentary irregularities); in this eye, the best best-correct visual acuity was 20/50 (Snellen equivalent) (extracted and modified from [4]).

Figure 4.

Figure 4

Advanced forms of age-related macular degeneration. (a) Autofluorescent retinography and (b) fluorescein angiography of two different cases of severe central geographic atrophy; in these eyes, the best-correct visual acuities were, respectively, 20/125 and 20/160 (Snellen equivalent). (c) Early and (d) late fluorescein angiograms of two different cases of subfoveal choroidal neovascularization; in both these eyes, the best-correct visual acuity was 20/200 (Snellen equivalent).

3. Complement System and Age-Related Macular Degeneration

Complement system consists of over 40 proteins and regulators which are detectable in the blood circulation. It plays a key role in host defense against pathogens, adaptive immune responses, removal of the immune complexes and apoptotic cells [79]. In humans, three complement-mediated pathways complementarily act, and each of them is characterized by a specific trigger as follows:

  1. antibody-antigen complex for the classical pathway;

  2. binding to host cell or pathogen surface for the alternative pathway;

  3. polysaccharides on microbial surfaces for the lectin pathway.

Dysregulation and/or dysfunction of the complement pathways can result in various critical autologous damages, with consequent pathogenetic implications in a wide spectrum of diseases [52]. Both pathogenesis and progression of AMD represent complex events, in which complement system is directly or indirectly implicated. Pathobiologic studies have identified numerous complement proteins inside drusen (i.e., the elementary clinical lesions identifiable in the macula of AMD patients), and genetic analyses have discovered the existence of common or rare polymorphisms in several complement-related genes that significantly increase or reduce the risk for AMD late in life [49]. In fact, the phenotypic features of drusen (i.e., clinical pattern and time of onset) and the genotypic individual background for AMD seem to be mutually and closely intersected with each other, figuratively sharing, as lowest common denominator, the local dysregulation of the complement system in the NR-RPE-BM-CC complex due to acquired and/or inherited risk factors. In particular, the development of AMD-related drusen occurs between the basal surface of RPE and the BM, a single-stratified extracellular matrix in contact with CC (Figures 2(a) and 3(a) [4]) [80]. Since the mid 1990s, increasing experimental and clinical evidences clearly indicate that a lot of complement-related molecules, such as complement activators, complement components, and complement regulatory proteins, represent substantial constituents of the drusen [35, 36, 65, 66, 73, 8193]. Starting from the beginning of the 2000s, the more and more exact identification of their compositional profile has been essential to create the basis for a new paradigm of AMD pathogenesis, in which macular and perimacular drusen should be considered as the earliest diagnosable byproducts of chronic local para-inflammatory phenomena at BM level. According to this model of AMD occurrence and progression, parainflammation of retinochoroidal tissues, accompanied by complement activation, immune-mediated processes, and bystander cell lysis, becomes the most crucial aspect of this neurodegenerative maculopathy [4752, 73, 85, 94].

In the course of the past few years, a definitive support for the “immuno-inflammatory” model of AMD pathogenesis has been evidenced by the clinical-genetic findings of numerous studies, which revealed highly significant correlations between AMD and polymorphisms of genes encoding for several molecules directly involved in the activities of the complement alternative pathway. Of these genes/loci, the most studied ones are:

  1. complement factor H (CFH) [1618, 9599],

  2. complement component 3 (C3) [100103],

  3. complement factor I (CFI) [104, 105],

  4. complement factor B (CFB) [19, 106, 107],

  5. complement component 2 (C2) [19, 106, 107],

  6. CFH-related genes (CFHR) type 1–5 [108110].

Even if some of these relationships between these genes/loci and AMD are incompletely understood, their comprehensive consideration indicates, once again, that complement-related polymorphic alleles are able to increase (CFH, C3, CFI, and CFHR-2-4-5) or reduce (CFB, C2, and CFHR-1-3) AMD risk, representing a central key point on which the evidences of the high heritability of AMD are based [23, 106, 111]. Then again, genetic susceptibility to AMD is a very multifaceted issue that also includes several other immunological/inflammatory aspects, either just indirectly linked or not linked to complement system such as, for example:

  1. interactions between C-reactive protein (CRP) and Y402H variant of CFH gene (rs1061170), a very common single nucleotide polymorphism (SNP) located within the chromosome 1q32 region and unequivocally identified in association with AMD among multiple study populations—providing for the first time a logical basis by which to assess the disease's risk in over 50% of affected individuals [49, 50, 52, 112];

  2. potential synergisms between the above-mentioned SNPs in genes/loci encoding for factors or components of the alternative complement pathway and some noncomplement-related genes, located on the chromosome 10q26 region and extensively described as strongly implicated in AMD pathogenesis, that is, the rs10490924 SNP of the age-related maculopathy susceptibility 2 (ARMS2), and the rs11200638 SNP of the high-temperature requirement factor A of serine peptidase 1 (HTRA1) [21, 22, 26, 49, 111].

CRP is a biomarker of acute-phase inflammation. It plays an essential role in the innate immune response to tissue injury and/or infection. Because CRP induces complement activation via the alternative pathway, it is plausible that CRP may have a direct responsibility in AMD pathogenesis by causing macular damages via complement-mediated mechanisms, as also happens in the case of CFH [46, 50, 71, 113, 114]. In fact, although several facets of the CFH-CRP interaction are not yet well defined [115], several findings have indicated that in carriers of the polymorphic H402 variant of CFH gene a lower affinity for CRP exists in respect of the individuals with the Y402 protein [92, 116, 117]. Moreover, a more recent study has confirmed that native CRP-CFH interaction is evident at high plasma CRP concentrations (as happens during the acute-phase response, i.e., when the H402 protein inadequately binds to CRP) [118], and also a large meta-analysis has documented that serum levels of CRP >3 mg/L are related to a double AMD risk in comparison with CRP concentrations <1 mg/L [119]. Starting from these latter results, it is not surprising that homozygous CFH-Y402H polymorphic genotype, together with elevated serum/plasma CRP levels, leads to a very high risk of both AMD and its progression (with odds ratios of 19.3 and 6.8, resp.) [120], even if the CRP elevation is not related to any variant of the CRP gene and no polymorphism in this gene is directly associated with AMD [121, 122]. As well, at the levels of RPE and choroid of CFH-H402 homozygous carriers, a greater amount of CRP was detected in comparison with that found in Y402 homozygotes, but there was no significant difference in CFH protein concentrations among individuals with diverse Y402H genotypes. This lack of local CRP expression indicates that CRP is present in the posterior segment of the eye as a consequence of deposition through chronic low-grade local inflammation [91].

Based on early genome-wide linkage analyses, which have established that the 10q26 locus is closely associated with AMD [123126], several clinical-genetic studies, specifically focused on this chromosomal region, discovered two major hereditary predisposing factors for AMD: the ARMS2 [127, 128] and the HTRA1 genes [129, 130]. At present, ARMS2 locus is considered a noncomplement-related gene because its potential role in the inflammatory process, if any, remains to be clarified [49]. In fact, although rs10490924-ARMS2 mRNA is detected in the human retina, both prevalent expression and cellular location of its putative protein are still under debate, having been initially observed in the mitochondrial outer membrane [131], and later in the cytosol and extracellular compartment [132, 133]. In any case, it seems extremely unlikely that deficiency of ARMS2-related protein could be a direct pathogenic mechanism responsible for AMD [134]. Also HTRA1 locus, encoding for a secreted protein belonging to the high-temperature requirement A family of serine proteases, can be still labeled as noncomplement-related gene [49]. However, because some molecules involved in the complement activities (i.e., clusterin, vitronectin, and fibromodulin) represent specific substrates for HTRA1 serine protease, an implication of HTRA1 in complement system has been notionally indicated [135]. The initial investigations documented the correlation between the rs11200638 promoter variant of the HTRA1 gene and an increased expression of its protein [129, 130], whereas other studies have not replicated these outcomes [131, 136]. Nevertheless, more recent reports showed that HTRA1 mRNA expression is higher in cultured RPE cells homozygous for the HTRA1 allele related to AMD risk, also supporting the perception that HTRA1 could be one of the causal genes in AMD patients [135, 137, 138].

In consideration of the heterogeneous gene-gene relationships between the major risk variants of the CFH, ARMS2, and HTRA1 loci, several Authors have emphasized the consistent possibility of an independent multiplicative joint effect in AMD, also taking into account that each of them should be contextualized within gene-environment interactions and epigenetic aspects [2225, 49, 139, 140]. Exclusively limiting the focus on those well-recognized SNPs which confer increased or decreased risk of inflammation (i.e., CFH, CX3CR1, IL-8, and TLR3 and 4), and voluntarily ignoring the other, suspected or ascertained, AMD-related gene variants (i.e., APOE, ABCR, LIPC, TIMP3, PON1, ERCC6, ELOVL4, fibulin-5, hemicentin-1, SERPING1, VLDLR, LRP6, VEGF, and KDR), the etiopathogenetic scenario of AMD is exactly that of a complex/polygenic disease characterized by (i) multiple clinical phenotypes with non-Mendelian transmission; (ii) environmental effects; (iii) increased incidence with age; (iv) specific susceptibility genes with variant alleles (Table 1) [20, 22, 25, 26, 29, 46, 140162]. The next section of this paper focuses on those immunological/inflammatory topics more directly translatable to improve the therapeutic strategies against AMD and, in particular, against its neovascular form, often responsible for the cases of most severe visual loss.

Table 1.

Main AMD-susceptibility genetic loci.

Locus Role in immunoinflammatory pathways Possibility of AMD-risk elevation
CFH Yes (complement system) High (in carriers of polymorphic allele)
ARMS2 Not clarified High (in carriers of polymorphic allele)
HTRA1 Possible (complement system) High (in carriers of polymorphic allele)
CFB Yes (complement system) Intermediate (in carriers of wild allele)
C2 Yes (complement system) Intermediate (in carriers of wild allele)
C3 Yes (complement system) Intermediate (in carriers of polymorphic allele)
CFI Yes (complement system) Low (in carriers of polymorphic allele)
TIMP3 Yes (immunity in extracellular matrix) Low (in carriers of polymorphic allele)
LIPC Not clarified Low (in carriers of polymorphic allele)
ABCR No Low (in carriers of polymorphic allele)
APOE No Low (in carriers of polymorphic allele)

Legend: CFH: complement factor H; ARMS2: age-related maculopathy susceptibility 2; HTRA1: high-temperature requirement factor A of serine peptidase 1; CFB: complement factor B; C2: complement component 2; C3: complement component 3; CFI: complement factor I; TIMP3: tissue inhibitor of metalloproteinases 3; LIPC: hepatic lipase gene; ABCR: ATP-binding cassette transporter; APOE: apolipoprotein E.

4. Agents Directed against the Immune Response and Age-Related Macular Degeneration

The responses of human immune system are necessary to defend our organism against several diseases, external antigens, invading microorganisms and/or acute tissue injuries. However, the contribution of the immune system in the occurrence of chronic age-related pathologic conditions has not been yet fully understood. In the course of the normal aging, as well as during chronic diseases, low-grade tissue stress (caused by noninfectious insults) may be related to subclinical damages resulting in the release of endogenous molecules, collectively called “alarmins”, that activate immunocompetent cells capable to support both innate and acquired immunity. In fact, they recruit and/or trigger receptor-expressing cells of the innate immune system, such as dendritic cells and macrophages, and consequently can also promote adaptive immunity in either direct or indirect manner [47]. To restore tissue homeostasis, by means of a mounting localized para-inflammatory response, immune system must be able to early identify such minimal bio-pathological changes in each specific district. Conversely, dysregulation or dysfunction of the immune system in chronically facing low-grade stress conditions may lead to manifest pathologies.

The retina, like the brain, is a high-metabolism tissue and is sensitive to noxious microenvironmental stimulations; however, unlike the brain, it is constantly exposed to the light, which can produce loads of photooxidized materials. Light-related stress and other oxidative damages increase in retinal tissue with the aging, as does the para-inflammatory response. In this view, although AMD is not a classic inflammatory disease, innate immunity and autoimmune components (i.e., complement factors, chemokines, cytokines, macrophages, and ocular microglia) have a reliable role in both pathogenesis and progression of AMD [47, 49, 163]. During the last two decades, directly or indirectly targeting these specific molecules/components, implicated in the immunoinflammatory pathways, has been assessed in the attempt to improve the therapeutic management of patients affected by the different clinical forms of AMD (Figures 24).

Since the mid 2000s, the Age-Related Eye Disease Study (AREDS, a large multicenter randomized clinical trial evaluating the long-term effects of high-dose antioxidant nutritional supplements on the incidence and progression of AMD and cataract) has documented a significantly lower incidence of advanced AMD in patients with drusen maculopathy treated with appropriate dosages of antioxidants than in a placebo group [164169]. Also other studies and evidences indicate the opportunity to indirectly counteract para-inflammatory changes minimizing the retinal oxidative stresses in AMD patients [11, 1315, 170, 171]. In particular, oral lutein intake results in beneficial effects on various visual function tests, and recent findings show that it is able to influence immune/inflammatory responses, not only diminishing the manifestation of various ocular inflammation models, but also suppressing NF kappa-B activation and/or inhibiting the expression of iNOS and COX-2 [48, 171].

On the other hand, during the last few years, numerous trials have been started to verify the therapeutic effects of various drugs aimed to directly downgrade the retinochoroidal immune response in AMD patients. In the next future, the outcomes of these ongoing clinical studies (156 studies found at http://clinicaltrials.gov/ searching, on June, 26 2012, with the keywords “age-related macular degeneration” and “anti-inflammatory”) together with the already reported findings [163, 172175] will be able to provide a more exact delineation of the role of the agents directed against the immune response in therapeutic recommendations for AMD patients [165168, 176186]. The majority of these interventional trials are conducted on patients affected by neovascular AMD, employing corticosteroids (i.e., dexamethasone and triamcinolone acetonide), nonsteroidal anti-inflammatory drugs (i.e., low-dose acetylsalicylic acid, bromfenac, diclofenac, and nepafenac), immunosuppressive agents (i.e., methotrexate and rapamycin), and biologics (i.e., anti-TNF-α agents such as infliximab and adalimumab, IL-2-receptor antagonists such as daclizumab, and complement inhibitors/regulators such as ARC1905, TNX-234, eculizumab, and POT-4), with the exception of rapamycin which has been also evaluated in cases of geographic atrophy secondary to AMD [163, 187].

The pathogenetic scenario that gives rise to the first RPE-BM-CC alterations in AMD is extremely complex. It includes a variety of predisposing genetic backgrounds, which can take effect on an heterogeneous plethora of para-inflammatory causative factors: cigarette smoking, phototoxic oxidative exposure, dietary habits, alterations of iron and lipid homeostasis, buildup of advanced glycation endproducts, microbial infection, lipofuscin and beta-amyloid toxicity, excessive immune-complex generation, choroidal hemodynamic insufficiency and ischemia, phagocytic overload, and/or RPE autophagy [49]. However, regardless of what are the conditions that can initially trigger the macular degenerative pattern in each individual AMD patient, it is indisputable that the decisive downstream consequences are the deposition and/or sequestration of both cellular and acellular debris at sub-RPE level. In the course of the normal human aging, mid- or long-term malfunctions in the tissue processing of these debris can be sufficient to locally generate abnormal para-inflammatory signal with a consequent aberrant activation of the complement system. This macular status would most likely result in persistent complement attack, further sub-RPE deposits, continuous formation of drusen, bystander injury to neighboring cells and, finally, irreversible photoreceptor degeneration and/or deconstruction (especially in those lots of adult or elder individuals who are more genetically susceptible to AMD for the presence of polymorphisms influencing the immune-inflammatory pathways and, in particular, the alternative complement-modulating activity) [25, 47, 49]. Starting from this rationale, our current knowledge regarding the role of both inflammation and complement systems in AMD should be refined to the point where it can be more easily translated in an innovative enhancement of AMD treatments, by means of either comparative randomized clinical trials or interventional pilot studies or biogenetic therapeutic researches.

5. Final Remarks

In the recent years, a substantial amount of evidences and/or arguments document the crucial responsibility of immune-inflammatory processes in the pathogenesis of AMD [3, 4752, 71, 188], clearly indicating the importance not only of specific complement-modulation agents, but also of nonspecific anti-inflammatory drugs, as adjunctive therapies for both non neovascular AMD (conventionally treated with AREDS formula and lutein) [163, 165171] and, most of all, neovascular AMD (routinely treated with intravitreal administration of drugs acting against vascular endothelial growth factor (anti-VEGF) and/or with photodynamic therapy with verteporfin (PDT-V)) [163, 172186, 189194]. The fact that para-inflammatory dysregulation is already present in the early stage of AMD may notionally support the preventive employment of agents directed against the immune-inflammatory response in combination with high-dose nutritional supplements (particularly in those patients with a disabling form of maculopathy in one eye, younger than 65 years, and/or carrier of significant genetic susceptibility to AMD) [30, 164171]. On the other hand, the existence of variable mid-term responsiveness of CNV to either anti-VEGF or PDT-V regimen (often resulting in elevated risks of legal blindness, high societal costs and expensive economic burden) practically recommends, above all in patients with advanced AMD in one eye, the adjunctive utilization of drugs directed against the immune-inflammatory response in combination with anti-VEGF injections and/or PDT-V [114, 171, 176179, 183185, 195217].

Returning to focus on the above-mentioned translational concepts about the opportunity of pharmacologic modulation toward the immune-inflammatory pathways in AMD, a comprehensive approach is warranted to verify the chances of a prompt application of this curative modality in the clinical setting. In theory, to modulate the complement attack and minimize the local parainflammation in AMD patients who carry one or more complement-related gene polymorphisms predisposing to the disease, the most specific approach would augment the retinochoroidal bioavailability of the native/protective form of those complement factors or components responsible for the genetic susceptibility to AMD [218220]. Adhering to this work hypothesis, a variety of delivery systems (i.e., gene transfer, cell-based therapies, organ (liver) transplantation, systemic or intraocular injections) can be envisioned to slow or arrest AMD by reasserting control over the complement system and, in particular, over its alternative pathway. If this biogenetic “augmentation” concept will be applicable also in the clinical AMD patterns, new complement-modulation therapeutics could be added to those several drugs directed against the immune-inflammatory response and already being tested on humans [49, 163]. However, at the moment, pending the concrete applicative possibilities of these biogenetic and/or pharmacologic complement-targeted treatments, open-label clinical trials are recommend, especially in patients with neovascular AMD, to better evaluate the therapeutic anti-CNV rationale in combining intravitreal corticosteroids either with the conventional anti-VEGF regimens or with anti-VEGF plus PDT-V customized protocols. In this view of good postmarketing study practice, as additional anti-CNV treatment, a promising anti-inflammatory strategy is that which involves the use of drug delivery systems (i.e., nonbiodegradable insert or biodegradable implant), able to provide a sustained release of intravitreal corticosteroids (fluocinolone acetonide or dexamethasone) for several months [221, 222]. In fact, taking into account both that immunoinflammatory phenomena are very active during the occurrence of an AMD-related CNV [71, 223], and that corticosteroids act upstream in immunoinflammatory cascades with consequent inhibition of the alternative-amplification of complement pathway [224, 225], and that a prolonged pharmacologic action represents an important parameter for the final efficacy of any therapy against neovascular AMD [176180], the above-described intraocular devices, already approved for the treatment of peculiar forms of macular edema and of noninfectious posterior uveitis [221, 222], could represent a rational adjunctive therapeutic approach for patients with neovascular AMD undergoing repeated intravitreal injections of anti-VEGF drug.

References

  • 1.Ambati J, Ambati BK, Yoo SH, Ianchulev S, Adamis AP. Age-related macular degeneration: etiology, pathogenesis, and therapeutic strategies. Survey of Ophthalmology. 2003;48(3):257–293. doi: 10.1016/s0039-6257(03)00030-4. [DOI] [PubMed] [Google Scholar]
  • 2.De Jong PTVM. Age-related macular degeneration. The New England Journal of Medicine. 2006;355(14):1474–1485. doi: 10.1056/NEJMra062326. [DOI] [PubMed] [Google Scholar]
  • 3.Coleman HR, Chan CC, Ferris FL, Chew EY. Age-related macular degeneration. The Lancet. 2008;372(9652):1835–1845. doi: 10.1016/S0140-6736(08)61759-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Saitta A. Anti-angiogenic drugs in the treatment of the neovascular age-related macular degeneration [M.S. thesis] The School of Medicine of the University of Ferrara; 2009. [Google Scholar]
  • 5.Resnikoff S, Pascolini D, Etya’ale D, et al. Global data on visual impairment in the year 2002. Bulletin of the World Health Organization. 2004;82(11):844–851. [PMC free article] [PubMed] [Google Scholar]
  • 6.Klein R, Cruickshanks KJ, Nash SD, et al. The prevalence of age-related macular degeneration and associated risk factors. Archives of Ophthalmology. 2010;128(6):750–758. doi: 10.1001/archophthalmol.2010.92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Friedman DS, O’Colmain BJ, Muñoz B, et al. Prevalence of age-related macular degeneration in the United States. Archives of Ophthalmology. 2004;122(4):564–572. doi: 10.1001/archopht.122.4.564. [DOI] [PubMed] [Google Scholar]
  • 8.Thornton J, Edwards R, Mitchell P, Harrison RA, Buchan I, Kelly SP. Smoking and age-related macular degeneration: a review of association. Eye. 2005;19(9):935–944. doi: 10.1038/sj.eye.6701978. [DOI] [PubMed] [Google Scholar]
  • 9.Chakravarthy U, Augood C, Bentham GC, et al. Cigarette smoking and age-related macular degeneration in the EUREYE Study. Ophthalmology. 2007;114(6):1157–1163. doi: 10.1016/j.ophtha.2006.09.022. [DOI] [PubMed] [Google Scholar]
  • 10.Kaushik S, Wang JJ, Flood V, et al. Dietary glycemic index and the risk of age-related macular degeneration. American Journal of Clinical Nutrition. 2008;88(4):1104–1110. doi: 10.1093/ajcn/88.4.1104. [DOI] [PubMed] [Google Scholar]
  • 11.Tan JSL, Wang JJ, Flood V, Rochtchina E, Smith W, Mitchell P. Dietary antioxidants and the long-term incidence of age-related macular degeneration: the Blue Mountains Eye Study. Ophthalmology. 2008;115(2):334–341. doi: 10.1016/j.ophtha.2007.03.083. [DOI] [PubMed] [Google Scholar]
  • 12.Kishan AU, Modjtahedi BS, Martins EN, Modjtahedi SP, Morse LS. Lipids and age-related macular degeneration. Survey of Ophthalmology. 2011;56(3):195–213. doi: 10.1016/j.survophthal.2010.08.008. [DOI] [PubMed] [Google Scholar]
  • 13.Wu J, Seregard S, Algvere PV. Photochemical damage of the retina. Survey of Ophthalmology. 2006;51(5):461–481. doi: 10.1016/j.survophthal.2006.06.009. [DOI] [PubMed] [Google Scholar]
  • 14.Wenzel A, Grimm C, Samardzija M, Remé CE. Molecular mechanisms of light-induced photoreceptor apoptosis and neuroprotection for retinal degeneration. Progress in Retinal and Eye Research. 2005;24(2):275–306. doi: 10.1016/j.preteyeres.2004.08.002. [DOI] [PubMed] [Google Scholar]
  • 15.Algvere PV, Marshall J, Seregard S. Age-related maculopathy and the impact of blue light hazard. Acta Ophthalmologica Scandinavica. 2006;84(1):4–15. doi: 10.1111/j.1600-0420.2005.00627.x. [DOI] [PubMed] [Google Scholar]
  • 16.Klein RJ, Zeiss C, Chew EY, et al. Complement factor H polymorphism in age-related macular degeneration. Science. 2005;308(5720):385–389. doi: 10.1126/science.1109557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Haines JL, Hauser MA, Schmidt S, et al. Complement factor H variant increases the risk of age-related macular degeneration. Science. 2005;308(5720):419–421. doi: 10.1126/science.1110359. [DOI] [PubMed] [Google Scholar]
  • 18.Hageman GS, Anderson DH, Johnson LV, et al. A common haplotype in the complement regulatory gene factor H (HF1/CFH) predisposes individuals to age-related macular degeneration. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(20):7227–7232. doi: 10.1073/pnas.0501536102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Spencer KL, Hauser MA, Olson LM, et al. Protective effect of complement factor B and complement component 2 variants in age-related macular degeneration. Human Molecular Genetics. 2007;16(16):1986–1992. [Google Scholar]
  • 20.Tuo J, Smith BC, Bojanowski CM, et al. The involvement of sequence variation and expression of CX3CR1 in the pathogenesis of age-related macular degeneration. The FASEB Journal. 2004;18(11):1297–1299. doi: 10.1096/fj.04-1862fje. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Bergeron-Sawitzke J, Gold B, Olsh A, et al. Multilocus analysis of age-related macular degeneration. European Journal of Human Genetics. 2009;17(9):1190–1199. doi: 10.1038/ejhg.2009.23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Leveziel N, Tilleul J, Puche N, et al. Genetic factors associated with age-related macular degeneration. Ophthalmologica. 2011;226(3):87–102. doi: 10.1159/000328981. [DOI] [PubMed] [Google Scholar]
  • 23.Seddon JM, Cote J, Page WF, Aggen SH, Neale MC. The US twin study of age-related macular degeneration: relative roles of genetic and environmental influences. Archives of Ophthalmology. 2005;123(3):321–327. doi: 10.1001/archopht.123.3.321. [DOI] [PubMed] [Google Scholar]
  • 24.Seddon JM, Reynolds R, Shah HR, Rosner B. Smoking, dietary betaine, methionine, and vitamin D in monozygotic twins with discordant macular degeneration: epigenetic implications. Ophthalmology. 2011;118(7):1386–1394. doi: 10.1016/j.ophtha.2010.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Hjelmeland LM. Dark matters in AMD genetics: epigenetics and stochasticity. Investigative Ophthalmology and Visual Science. 2011;52(3):1622–1631. doi: 10.1167/iovs.10-6765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Tuo J, Grob S, Zhang K, Chan CC. Genetics of immunological and inflammatory components in age-related macular degeneration. Ocular Immunology and Inflammation. 2012;20(1):27–36. doi: 10.3109/09273948.2011.628432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Rienhoff HY. Genomewide association studies and assessment of risk of disease. The New England Journal of Medicine. 2010;363(21):p. 2077. doi: 10.1056/NEJMc1010310. [DOI] [PubMed] [Google Scholar]
  • 28.Peter I, Seddon JM. Genetic epidemiology: successes and challenges of genome-wide association studies using the example of age-related macular degeneration. American Journal of Ophthalmology. 2010;150(4):450–452. doi: 10.1016/j.ajo.2010.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Francis PJ, Hamon SC, Ott J, Weleber RG, Klein ML. Polymorphisms in C2, CFB and C3 are associated with progression to advanced age related macular degeneration associated with visual loss. Journal of Medical Genetics. 2009;46(5):300–307. doi: 10.1136/jmg.2008.062737. [DOI] [PubMed] [Google Scholar]
  • 30.Ricci F, Zampatti S, D’Abbruzzi F, et al. Typing of ARMS2 and CFH in age-related macular degeneration: case-control study and assessment of frequency in the Italian population. Archives of Ophthalmology. 2009;127(10):1368–1372. doi: 10.1001/archophthalmol.2009.237. [DOI] [PubMed] [Google Scholar]
  • 31.Ryu E, Fridley BL, Tosakulwong N, Bailey KR, Edwards AO. Genome-wide association analyses of genetic, phenotypic, and environmental risks in the age-related eye disease study. Molecular Vision. 2010;16:2811–2821. [PMC free article] [PubMed] [Google Scholar]
  • 32.Meyer KJ, Davis LK, Schindler EI, et al. Genome-wide analysis of copy number variants in age-related macular degeneration. Human Genetics. 2011;129(1):91–100. doi: 10.1007/s00439-010-0904-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Penfold PL, Killingsworth MC, Sarks SH. Senile macular degeneration: the involvement of immunocompetent cells. Graefe’s Archive for Clinical and Experimental Ophthalmology. 1985;223(2):69–76. doi: 10.1007/BF02150948. [DOI] [PubMed] [Google Scholar]
  • 34.Lopez PF, Grossniklaus HE, Lambert HM, et al. Pathologic features of surgically excised subretinal neovascular membranes in age-related macular degeneration. American Journal of Ophthalmology. 1991;112(6):647–656. doi: 10.1016/s0002-9394(14)77270-8. [DOI] [PubMed] [Google Scholar]
  • 35.Mullins RF, Russell SR, Anderson DH, Hageman GS. Drusen associated with aging and age-related macular degeneration contain proteins common to extracellular deposits associated with atherosclerosis, elastosis, amyloidosis, and dense deposit disease. The FASEB Journal. 2000;14(7):835–846. [PubMed] [Google Scholar]
  • 36.Johnson LV, Ozaki S, Staples MK, Erickson PA, Anderson DH. A potential role for immune complex pathogenesis in drusen formation. Experimental Eye Research. 2000;70(4):441–449. doi: 10.1006/exer.1999.0798. [DOI] [PubMed] [Google Scholar]
  • 37.Nozaki M, Raisler BJ, Sakurai E, et al. Drusen complement components C3a and C5a promote choroidal neovascularization. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(7):2328–2333. doi: 10.1073/pnas.0408835103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Lommatzsch A, Hermans P, Weber B, Pauleikhoff D. Complement factor H variant Y402H and basal laminar deposits in exudative age-related macular degeneration. Graefe’s Archive for Clinical and Experimental Ophthalmology. 2007;245(11):1713–1716. doi: 10.1007/s00417-007-0649-7. [DOI] [PubMed] [Google Scholar]
  • 39.Lommatzsch A, Hermans P, Müller KD, Bornfeld N, Bird AC, Pauleikhoff D. Are low inflammatory reactions involved in exudative age-related macular degeneration? Graefe’s Archive for Clinical and Experimental Ophthalmology. 2008;246(6):803–810. doi: 10.1007/s00417-007-0749-4. [DOI] [PubMed] [Google Scholar]
  • 40.Bhutto IA, Baba T, Merges C, Juriasinghani V, McLeod DS, Lutty GA. C-reactive protein and complement factor H in aged human eyes and eyes with age-related macular degeneration. British Journal of Ophthalmology. 2011;95(9):1323–1330. doi: 10.1136/bjo.2010.199216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Fett AL, Hermann MM, Muether PS, Kirchhof B, Fauser S. Immunohistochemical localization of complement regulatory proteins in the human retina. Histology and Histopathology. 2012;27(3):357–364. doi: 10.14670/HH-27.357. [DOI] [PubMed] [Google Scholar]
  • 42.Sivaprasad S, Adewoyin T, Bailey TA, et al. Estimation of systemic complement C3 activity in age-related macular degeneration. Archives of Ophthalmology. 2007;125(4):515–519. doi: 10.1001/archopht.125.4.515. [DOI] [PubMed] [Google Scholar]
  • 43.Scholl HPN, Issa PC, Walier M, et al. Systemic complement activation in age-related macular degeneration. PLoS ONE. 2008;3(7) doi: 10.1371/journal.pone.0002593.e2593 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.MacHalińska A, Dziedziejko V, Mozolewska-Piotrowska K, Karczewicz D, Wiszniewska B, MacHaliński B. Elevated plasma levels of c3a complement compound in the exudative form of age-related macular degeneration. Ophthalmic Research. 2009;42(1):54–59. doi: 10.1159/000219686. [DOI] [PubMed] [Google Scholar]
  • 45.Reynolds R, Hartnett ME, Atkinson JP, Giclas PC, Rosner B, Seddon JM. Plasma complement components and activation fragments: associations with age-related macular degeneration genotypes and phenotypes. Investigative Ophthalmology and Visual Science. 2009;50(12):5818–5827. doi: 10.1167/iovs.09-3928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ding X, Patel M, Chan CC. Molecular pathology of age-related macular degeneration. Progress in Retinal and Eye Research. 2009;28(1):1–18. doi: 10.1016/j.preteyeres.2008.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Xu H, Chen M, Forrester JV. Para-inflammation in the aging retina. Progress in Retinal and Eye Research. 2009;28(5):348–368. doi: 10.1016/j.preteyeres.2009.06.001. [DOI] [PubMed] [Google Scholar]
  • 48.Chen M, Forrester JV, Xu H. Dysregulation in retinal para-inflammation and age-related retinal degeneration in CCL2 or CCR2 deficient mice. PLoS ONE. 2011;6(8) doi: 10.1371/journal.pone.0022818.e22818 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Anderson DH, Radeke MJ, Gallo NB, et al. The pivotal role of the complement system in aging and age-related macular degeneration: hypothesis re-visited. Progress in Retinal and Eye Research. 2010;29(2):95–112. doi: 10.1016/j.preteyeres.2009.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Donoso LA, Vrabec T, Kuivaniemi H. The role of complement factor H in age-related macular degeneration: a review. Survey of Ophthalmology. 2010;55(3):227–246. doi: 10.1016/j.survophthal.2009.11.001. [DOI] [PubMed] [Google Scholar]
  • 51.Johnson LV, Forest DL, Banna CD, et al. Cell culture model that mimics drusen formation and triggers complement activation associated with age-related macular degeneration. Proceedings of the National Academy of Sciences of USA. 2011;108(45):18277–18282. doi: 10.1073/pnas.1109703108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Khandhadia S, Cipriani V, Yates JR, Lotery AJ. Age-related macular degeneration and the complement system. Immunobiology. 2012;217(2):127–146. doi: 10.1016/j.imbio.2011.07.019. [DOI] [PubMed] [Google Scholar]
  • 53.Medzhitov R. Origin and physiological roles of inflammation. Nature. 2008;454(7203):428–435. doi: 10.1038/nature07201. [DOI] [PubMed] [Google Scholar]
  • 54.Harman D. Aging: a theory based on free radical and radiation chemistry. Journal of gerontology. 1956;11(3):298–300. doi: 10.1093/geronj/11.3.298. [DOI] [PubMed] [Google Scholar]
  • 55.Seddon JM, George S, Rosner B, Rifai N. Progression of age-related macular degeneration: prospective assessment of C-reactive protein, interleukin 6, and other cardiovascular biomarkers. Archives of Ophthalmology. 2005;123(6):774–782. doi: 10.1001/archopht.123.6.774. [DOI] [PubMed] [Google Scholar]
  • 56.Wei YH, Lu CY, Lee HC, Pang CY, Ma YS. Oxidative damage and mutation to mitochondrial DNA and age-dependent decline of mitochondrial respiratory function. Annals of the New York Academy of Sciences. 1998;854:155–170. doi: 10.1111/j.1749-6632.1998.tb09899.x. [DOI] [PubMed] [Google Scholar]
  • 57.Hekimi S, Lapointe J, Wen Y. Taking a "good" look at free radicals in the aging process. Trends in Cell Biology. 2011;21(10):569–576. doi: 10.1016/j.tcb.2011.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Melo A, Monteiro L, Lima RM, Oliveira DM, Cerqueira MD, El-Bachá RS. Oxidative stress in neurodegenerative diseases: mechanisms and therapeutic perspectives. Oxidative Medicine and Cellular Longevity. 2011;2011:14 pages. doi: 10.1155/2011/467180.467180 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Garbin U, Pasini AF, Stranieri C, et al. Cigarette smoking blocks the protective expression of Nrf2/ARE pathway in peripheral mononuclear cells of young heavy smokers favouring inflammation. PLoS ONE. 2009;4(12) doi: 10.1371/journal.pone.0008225.e8225 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Calder PC, Ahluwalia N, Brouns F, et al. Dietary factors and low-grade inflammation in relation to overweight and obesity. British Journal of Nutrition. 2011;106(3):5–78. doi: 10.1017/S0007114511005460. [DOI] [PubMed] [Google Scholar]
  • 61.Heck DE, Vetrano AM, Mariano TM, Laskin JD. UVB light stimulates production of reactive oxygen species: unexpected role for catalase. The Journal of Biological Chemistry. 2003;278(25):22432–22436. doi: 10.1074/jbc.C300048200. [DOI] [PubMed] [Google Scholar]
  • 62.Naik E, Dixit VM. Mitochondrial reactive oxygen species drive proinflammatory cytokine production. Journal of Experimental Medicine. 2011;208(3):417–420. doi: 10.1084/jem.20110367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Burton GJ. Oxygen, the Janus gas; its effects on human placental development and function. Journal of Anatomy. 2009;215(1):27–35. doi: 10.1111/j.1469-7580.2008.00978.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Kajimoto H, Hashimoto K, Bonnet SN, et al. Oxygen activates the rho/rho-kinase pathway and induces RhoB and ROCK-1 expression in human and rabbit ductus arteriosus by increasing mitochondria-derived reactive oxygen species: a newly recognized mechanism for sustaining ductal constriction. Circulation. 2007;115(13):1777–1788. doi: 10.1161/CIRCULATIONAHA.106.649566. [DOI] [PubMed] [Google Scholar]
  • 65.Zhou J, Jang YP, Kim SR, Sparrow JR. Complement activation by photooxidation products of A2E, a lipofuscin constituent of the retinal pigment epithelium. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(44):16182–16187. doi: 10.1073/pnas.0604255103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Zhou J, Kim SR, Westlund BS, Sparrow JR. Complement activation by bisretinoid constituents of RPE lipofuscin. Investigative Ophthalmology and Visual Science. 2009;50(3):1392–1399. doi: 10.1167/iovs.08-2868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Dunaief JL, Dentchev T, Ying GS, Milam AH. The role of apoptosis in age-related macular degeneration. Archives of Ophthalmology. 2002;120(11):1435–1442. doi: 10.1001/archopht.120.11.1435. [DOI] [PubMed] [Google Scholar]
  • 68.Takahashi A, Masuda A, Sun M, Centonze VE, Herman B. Oxidative stress-induced apoptosis is associated with alterations in mitochondrial caspase activity and Bcl-2-dependent alterations in mitochondrial pH (pHm) Brain Research Bulletin. 2004;62(6):497–504. doi: 10.1016/j.brainresbull.2003.07.009. [DOI] [PubMed] [Google Scholar]
  • 69.Ushio-Fukai M. Redox signaling in angiogenesis: role of NADPH oxidase. Cardiovascular Research. 2006;71(2):226–235. doi: 10.1016/j.cardiores.2006.04.015. [DOI] [PubMed] [Google Scholar]
  • 70.Ushio-Fukai M. VEGF signaling through NADPH oxidase-derived ROS. Antioxidants and Redox Signaling. 2007;9(6):731–739. doi: 10.1089/ars.2007.1556. [DOI] [PubMed] [Google Scholar]
  • 71.Campa C, Costagliola C, Incorvaia C, et al. Inflammatory mediators and angiogenic factors in choroidal neovascularization: pathogenetic interactions and therapeutic implications. Mediators of Inflammation. 2010;2010:14 pages. doi: 10.1155/2010/546826.546826 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Wang H, Wittchen ES, Hartnett ME. Breaking barriers: insight into the pathogenesis of neovascular age-related macular degeneration. Eye and Brain. 2011;3:19–28. doi: 10.2147/EB.S24951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Anderson DH, Mullins RF, Hageman GS, Johnson LV. A role for local inflammation in the formation of drusen in the aging eye. American Journal of Ophthalmology. 2002;134(3):411–431. doi: 10.1016/s0002-9394(02)01624-0. [DOI] [PubMed] [Google Scholar]
  • 74.Gao H, Hollyfield JG. Aging of the human retina: differential loss of neurons and retinal pigment epithelial cells. Investigative Ophthalmology and Visual Science. 1992;33(1):1–17. [PubMed] [Google Scholar]
  • 75.Curcio CA, Millican CL, Allen KA, Kalina RE. Aging of the human photoreceptor mosaic: evidence for selective vulnerability of rods in central retina. Investigative Ophthalmology and Visual Science. 1993;34(12):3278–3296. [PubMed] [Google Scholar]
  • 76.Newsome DA, Huh W, Green WR. Bruch’s membrane age-related changes vary by region. Current Eye Research. 1987;6(10):1211–1221. doi: 10.3109/02713688709025231. [DOI] [PubMed] [Google Scholar]
  • 77.Ramrattan RS, Van der Schaft TL, Mooy CM, De Bruijn WC, Mulder PGH, De Jong PTVM. Morphometric analysis of Bruch’s membrane, the choriocapillaris, and the choroid in aging. Investigative Ophthalmology and Visual Science. 1994;35(6):2857–2864. [PubMed] [Google Scholar]
  • 78.Xu H, Dawson R, Forrester JV, Liversidge J. Identification of novel dendritic cell populations in normal mouse retina. Investigative Ophthalmology and Visual Science. 2007;48(4):1701–1710. doi: 10.1167/iovs.06-0697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Walport MJ. Complement (first of two parts) The New England Journal of Medicine. 2001;344(14):1058–1066. doi: 10.1056/NEJM200104053441406. [DOI] [PubMed] [Google Scholar]
  • 80.Hogan MJ, Alvarado JA, Weddell JE. Histology of the Human Eye. WB Saunders Company; 1971. [Google Scholar]
  • 81.Van der Schaft TL, Mooy CM, De Bruijn WC, de Jong PTVM. Early stages of age-related macular degeneration: an immunofluorescence and electron microscopy study. British Journal of Ophthalmology. 1993;77(10):657–661. doi: 10.1136/bjo.77.10.657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Mullins RF, Hageman GS. Histochemical comparison of ocular “drusen” in monkey and human. In: La Vail MM, Hollyfield JG, Anderson RE, editors. Degenerative Retinal Diseases. Plenum; 1997. pp. 1–10. [Google Scholar]
  • 83.Hageman GS, Mullins RF, Russell SR, Johnson LV, Anderson DH. Vitronectin is a constituent of ocular drusen and the vitronectin gene is expressed in human retinal pigmented epithelial cells. The FASEB Journal. 1999;13(3):477–484. doi: 10.1096/fasebj.13.3.477. [DOI] [PubMed] [Google Scholar]
  • 84.Hageman GS, Mullins RF. Molecular composition of drusen as related to substructural phenotype. Molecular vision. 1999;5:p. 28. [PubMed] [Google Scholar]
  • 85.Johnson LV, Leitner WP, Staples MK, Anderson DH. Complement activation and inflammatory processes in drusen formation and age related macular degeneration. Experimental Eye Research. 2001;73(6):887–896. doi: 10.1006/exer.2001.1094. [DOI] [PubMed] [Google Scholar]
  • 86.Mullins RF, Aptsiauri N, Hageman GS. Structure and composition of drusen associated with glomerulonephritis: implications for the role of complement activation in drusen biogenesis. Eye. 2001;15(3):390–395. doi: 10.1038/eye.2001.142. [DOI] [PubMed] [Google Scholar]
  • 87.Johnson LV, Leitner WP, Rivest AJ, Staples MK, Radeke MJ, Anderson DH. The Alzheimer’s Aβ-peptide is deposited at sites of complement activation in pathologic deposits associated with aging and age-related macular degeneration. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(18):11830–11835. doi: 10.1073/pnas.192203399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Crabb JW, Miyagi M, Gu X, et al. Drusen proteome analysis: an approach to the etiology of age-related macular degeneration. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(23):14682–14687. doi: 10.1073/pnas.222551899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Sakaguchi H, Miyagi M, Shadrach KG, Rayborn ME, Crabb JW, Hollyfield JG. Clusterin is present in drusen in age-related macular degeneration. Experimental Eye Research. 2002;74(4):547–549. doi: 10.1006/exer.2002.1186. [DOI] [PubMed] [Google Scholar]
  • 90.Anderson DH, Talaga KC, Rivest AJ, Barron E, Hageman GS, Johnson LV. Characterization of β amyloid assemblies in drusen: the deposits associated with aging and age-related macular degeneration. Experimental Eye Research. 2004;78(2):243–256. doi: 10.1016/j.exer.2003.10.011. [DOI] [PubMed] [Google Scholar]
  • 91.Johnson PT, Betts KE, Radeke MJ, Hageman GS, Anderson DH, Johnson LV. Individuals homozygous for the age-related macular degeneration risk-conferring variant of complement factor H have elevated levels of CRP in the choroid. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(46):17456–17461. doi: 10.1073/pnas.0606234103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Laine M, Jarva H, Seitsonen S, et al. Y402H polymorphism of complement factor H affects binding affinity to C-reactive protein. Journal of Immunology. 2007;178(6):3831–3836. doi: 10.4049/jimmunol.178.6.3831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Wang J, Ohno-Matsui K, Yoshida T, et al. Altered function of factor I caused by amyloid β: implication for pathogenesis of age-related macular degeneration from drusen. Journal of Immunology. 2008;181(1):712–720. doi: 10.4049/jimmunol.181.1.712. [DOI] [PubMed] [Google Scholar]
  • 94.Hageman GS, Luthert PJ, Victor Chong NH, Johnson LV, Anderson DH, Mullins RF. An integrated hypothesis that considers drusen as biomarkers of immune-mediated processes at the RPE-Bruch’s membrane interface in aging and age-related macular degeneration. Progress in Retinal and Eye Research. 2001;20(6):705–732. doi: 10.1016/s1350-9462(01)00010-6. [DOI] [PubMed] [Google Scholar]
  • 95.Edwards AO, Ritter R, Abel KJ, Manning A, Panhuysen C, Farrer LA. Complement factor H polymorphism and age-related macular degeneration. Science. 2005;308(5720):421–424. doi: 10.1126/science.1110189. [DOI] [PubMed] [Google Scholar]
  • 96.Despriet DDG, Klaver CCW, Witteman JCM, et al. Complement factor H polymorphism, complement activators, and risk of age-related macular degeneration. Journal of the American Medical Association. 2006;296(3):301–309. doi: 10.1001/jama.296.3.301. [DOI] [PubMed] [Google Scholar]
  • 97.Li M, Atmaca-Sonmez P, Othman M, et al. CFH haplotypes without the Y402H coding variant show strong association with susceptibility to age-related macular degeneration. Nature Genetics. 2006;38(9):1049–1054. doi: 10.1038/ng1871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Francis PJ, Schultz DW, Hamon S, Ott J, Weleber RG, Klein ML. Haplotypes in the complement factor H (CFH) gene: associations with drusen and advanced age-related macular degeneration. PLoS ONE. 2007;2(11) doi: 10.1371/journal.pone.0001197.e1197 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Seddon JM, Francis PJ, George S, Schultz DW, Rosner B, Klein ML. Association of CFH Y402H and LOC387715 A69S with progression of age-related macular degeneration. Journal of the American Medical Association. 2007;297(16):1793–1800. doi: 10.1001/jama.297.16.1793. [DOI] [PubMed] [Google Scholar]
  • 100.Yates JRW, Sepp T, Matharu BK, et al. Complement C3 variant and the risk of age-related macular degeneration. The New England Journal of Medicine. 2007;357(6):553–561. doi: 10.1056/NEJMoa072618. [DOI] [PubMed] [Google Scholar]
  • 101.Maller JB, Fagerness JA, Reynolds RC, Neale BM, Daly MJ, Seddon JM. Variation in complement factor 3 is associated with risk of age-related macular degeneration. Nature Genetics. 2007;39(10):1200–1201. doi: 10.1038/ng2131. [DOI] [PubMed] [Google Scholar]
  • 102.Park KH, Fridley BL, Ryu E, Tosakulwong N, Edwards AO. Complement component 3 (C3) haplotypes and risk of advanced age-related macular degeneration. Investigative Ophthalmology and Visual Science. 2009;50(7):3386–3393. doi: 10.1167/iovs.08-3231. [DOI] [PubMed] [Google Scholar]
  • 103.Despriet DDG, van Duijn CM, Oostra BA, et al. Complement component C3 and risk of age-related macular degeneration. Ophthalmology. 2009;116(3):474–480. doi: 10.1016/j.ophtha.2008.09.055. [DOI] [PubMed] [Google Scholar]
  • 104.Fagerness JA, Maller JB, Neale BM, Reynolds RC, Daly MJ, Seddon JM. Variation near complement factor I is associated with risk of advanced AMD. European Journal of Human Genetics. 2009;17(1):100–104. doi: 10.1038/ejhg.2008.140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Kondo N, Bessho H, Honda S, Negi A. Additional evidence to support the role of a common variant near the complement factor i gene in susceptibility to age-related macular degeneration. European Journal of Human Genetics. 2010;18(6):634–635. doi: 10.1038/ejhg.2009.243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Gold B, Merriam JE, Zernant J, et al. Variation in factor B (BF) and complement component 2 (C2) genes is associated with age-related macular degeneration. Nature Genetics. 2006;38(4):458–462. doi: 10.1038/ng1750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Jakobsdottir J, Conley YP, Weeks DE, Ferrell RE, Gorin MB. C2 and CFB genes in age-related maculopathy and joint action with CFH and LOC387715 genes. PLoS ONE. 2008;3(5) doi: 10.1371/journal.pone.0002199.e2199 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Hageman GS, Hancox LS, Taiber AJ, et al. Extended haplotypes in the complement factor H (CFH) and CFH-related (CFHR) family of genes protect against age-related macular degeneration: characterization, ethnic distribution and evolutionary implications. Annals of Medicine. 2006;38(8):592–604. [PMC free article] [PubMed] [Google Scholar]
  • 109.Hughes AE, Orr N, Esfandiary H, Diaz-Torres M, Goodship T, Chakravarthy U. A common CFH haplotype, with deletion of CFHR1 and CFHR3, is associated with lower risk of age-related macular degeneration. Nature Genetics. 2006;38(10):1173–1177. doi: 10.1038/ng1890. [DOI] [PubMed] [Google Scholar]
  • 110.Zhang H, Morrison MA, DeWan A, et al. The NEI/NCBI dbGAP database: genotypes and haplotypes that may specifically predispose to risk of neovascular age-related macular degeneration. BMC Medical Genetics. 2008;9:p. 51. doi: 10.1186/1471-2350-9-51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Francis PJ, Klein ML. Update on the role of genetics in the onset of age-related macular degeneration. Journal of Clinical Ophthalmology. 2011;5:1127–1133. doi: 10.2147/OPTH.S11627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Seddon JM, Reynolds R, Maller J, Fagerness JA, Daly MJ, Rosner B. Prediction model for prevalence and incidence of advanced age-related macular degeneration based on genetic, demographic, and environmental variables. Investigative Ophthalmology and Visual Science. 2009;50(5):2044–2053. doi: 10.1167/iovs.08-3064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Du Clos TW. Function of C-reactive protein. Annals of Medicine. 2000;32(4):274–278. doi: 10.3109/07853890009011772. [DOI] [PubMed] [Google Scholar]
  • 114.Parmeggiani F, Gemmati D, Costagliola C, et al. Genetic predictors of response to photodynamic therapy. Molecular Diagnosis & Therapy. 2011;15(4):195–210. doi: 10.1007/BF03256411. [DOI] [PubMed] [Google Scholar]
  • 115.Hakobyan S, Harris CL, Van Den Berg CW, et al. Complement factor H binds to denatured rather than to native pentameric C-reactive protein. The Journal of Biological Chemistry. 2008;283(45):30451–30460. doi: 10.1074/jbc.M803648200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Herbert AP, Deakin JA, Schmidt CQ, et al. Structure shows that a glycosaminoglycan and protein recognition site in factor H is perturbed by age-related macular degeneration-linked single nucleotide polymorphism. The Journal of Biological Chemistry. 2007;282(26):18960–18968. doi: 10.1074/jbc.M609636200. [DOI] [PubMed] [Google Scholar]
  • 117.Yu J, Wiita P, Kawaguchi R, et al. Biochemical analysis of a common human polymorphism associated with age-related macular degeneration. Biochemistry. 2007;46(28):8451–8461. doi: 10.1021/bi700459a. [DOI] [PubMed] [Google Scholar]
  • 118.Perkins SJ, Nan R, Okemefuna AI, Li K, Khan S, Miller A. Multiple interactions of complement factor h with its ligands in solution: a progress report. Advances in Experimental Medicine and Biology. 2010;703:25–47. doi: 10.1007/978-1-4419-5635-4_3. [DOI] [PubMed] [Google Scholar]
  • 119.Hong T, Tan AG, Mitchell P, Wang JJ. A review and meta-analysis of the association between C-reactive protein and age-related macular degeneration. Survey of Ophthalmology. 2011;56(3):184–194. doi: 10.1016/j.survophthal.2010.08.007. [DOI] [PubMed] [Google Scholar]
  • 120.Robman L, Baird PN, Dimitrov PN, Richardson AJ, Guymer RH. C-reactive protein levels and complement factor H polymorphism interaction in age-related macular degeneration and its progression. Ophthalmology. 2010;117(10):1982–1988. doi: 10.1016/j.ophtha.2010.02.003. [DOI] [PubMed] [Google Scholar]
  • 121.Schaumberg DA, Christen WG, Kozlowski P, Miller DT, Ridker PM, Zee RYL. A prospective assessment of the Y402H variant in complement factor H, genetic variants in C-reactive protein, and risk of age-related macular degeneration. Investigative Ophthalmology and Visual Science. 2006;47(6):2336–2340. doi: 10.1167/iovs.05-1456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Kim IK, Ji F, Morrison MA, et al. Comprehensive analysis of CRP, CFH Y402H and environmental risk factors on risk of neovascular age-related macular degeneration. Molecular Vision. 2008;14:1487–1495. [PMC free article] [PubMed] [Google Scholar]
  • 123.Iyengar SK, Song D, Klein BEK, et al. Dissection of genomewide-scan data in extended families reveals a major locus and oligogenic susceptibility for age-related macular degeneration. American Journal of Human Genetics. 2004;74(1):20–39. doi: 10.1086/380912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Majewski J, Schultz DW, Weleber RG, et al. Age-related macular degeneration—a genome scan in extended families. American Journal of Human Genetics. 2003;73(3):540–550. doi: 10.1086/377701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Seddon JM, Santangelo SL, Book K, Chong S, Cote J. A genomewide scan for age-related macular degeneration provides evidence for linkage to several chromosomal regions. American Journal of Human Genetics. 2003;73(4):780–790. doi: 10.1086/378505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Weeks DE, Conley YP, Mah TS, et al. A full genome scan for age-related maculopathy. Human Molecular Genetics. 2000;9(9):1329–1349. doi: 10.1093/hmg/9.9.1329. [DOI] [PubMed] [Google Scholar]
  • 127.Jakobsdottir J, Conley YP, Weeks DE, Mah TS, Ferrell RE, Gorin MB. Susceptibility genes for age-related maculopathy on chromosome 10q26. American Journal of Human Genetics. 2005;77(3):389–407. doi: 10.1086/444437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Rivera A, Fisher SA, Fritsche LG, et al. Hypothetical LOC387715 is a second major susceptibility gene for age-related macular degeneration, contributing independently of complement factor H to disease risk. Human Molecular Genetics. 2005;14(21):3227–3236. doi: 10.1093/hmg/ddi353. [DOI] [PubMed] [Google Scholar]
  • 129.DeWan A, Liu M, Hartman S, et al. HTRA1 promoter polymorphism in wet age-related macular degeneration. Science. 2006;314(5801):989–992. doi: 10.1126/science.1133807. [DOI] [PubMed] [Google Scholar]
  • 130.Yang Z, Camp NJ, Sun H, et al. A variant of the HTRA1 gene increases susceptibility to age-related macular degeneration. Science. 2006;314(5801):992–993. doi: 10.1126/science.1133811. [DOI] [PubMed] [Google Scholar]
  • 131.Kanda A, Chen W, Othman M, et al. A variant of mitochondrial protein LOC387715/ARMS2, not HTRA1, is strongly associated with age-related macular degeneration. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(41):16227–16232. doi: 10.1073/pnas.0703933104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Wang G, Spencer KL, Court BL, et al. Localization of age-related macular degeneration-associated ARMS2 in cytosol, not mitochondria. Investigative Ophthalmology and Visual Science. 2009;50(7):3084–3090. doi: 10.1167/iovs.08-3240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Kortvely E, Hauck SM, Duetsch G, et al. ARMS2 is a constituent of the extracellular matrix providing a link between familial and sporadic age-related macular degenerations. Investigative Ophthalmology and Visual Science. 2010;51(1):79–88. doi: 10.1167/iovs.09-3850. [DOI] [PubMed] [Google Scholar]
  • 134.Friedrich U, Myers CA, Fritsche LG, et al. Risk-and non-risk-associated variants at the 10q26 AMD locus influence ARMS2 mRNA expression but exclude pathogenic effects due to protein deficiency. Human Molecular Genetics. 2011;20(7):1387–1399. doi: 10.1093/hmg/ddr020.ddr020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.An E, Sen S, Park SK, Gordish-Dressman H, Hathout Y. Identification of novel substrates for the serine protease HTRA1 in the human RPE secretome. Investigative Ophthalmology and Visual Science. 2010;51(7):3379–3386. doi: 10.1167/iovs.09-4853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Kanda A, Stambolian D, Chen W, Curcio CA, Abecasis GR, Swaroop A. Age-related macular degeneration-associated variants at chromosome 10q26 do not significantly alter ARMS2 and HTRA1 transcript levels in the human retina. Molecular Vision. 2010;16:1317–1323. [PMC free article] [PubMed] [Google Scholar]
  • 137.Yang Z, Tong Z, Chen Y, et al. Genetic and functional dissection of HTRA1 and LOC387715 in age-related macular degeneration. PLoS Genetics. 2010;6(2) doi: 10.1371/journal.pgen.1000836.e1000836 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Ng TK, Yam GH, Chen WQ, et al. Interactive expressions of HtrA1 and VEGF in human vitreous humors and fetal RPE cells. Investigative Ophthalmology & Visual Science. 2011;52(6):3706–3712. doi: 10.1167/iovs.10-6773. [DOI] [PubMed] [Google Scholar]
  • 139.Schmidt S, Hauser MA, Scott WK, et al. Cigarette smoking strongly modifies the association of LOC387715 and age-related macular degeneration. American Journal of Human Genetics. 2006;78(5):852–864. doi: 10.1086/503822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Conley YP, Jakobsdottir J, Mah T, et al. CFH, ELOVL4, PLEKHA1 and LOC387715 genes and susceptibility to age-related maculopathy: AREDS and CHS cohorts and meta-analyses. Human Molecular Genetics. 2006;15(21):3206–3218. doi: 10.1093/hmg/ddl396. [DOI] [PubMed] [Google Scholar]
  • 141.Tsai YY, Lin JM, Wan L, et al. Interleukin gene polymorphisms in age-related macular degeneration. Investigative Ophthalmology and Visual Science. 2008;49(2):693–698. doi: 10.1167/iovs.07-0125. [DOI] [PubMed] [Google Scholar]
  • 142.Zareparsi S, Buraczynska M, Branham KEH, et al. Toll-like receptor 4 variant D299G is associated with susceptibility to age-related macular degeneration. Human Molecular Genetics. 2005;14(11):1449–1455. doi: 10.1093/hmg/ddi154. [DOI] [PubMed] [Google Scholar]
  • 143.Yang Z, Stratton C, Francis PJ, et al. Toll-like receptor 3 and geographic atrophy in age-related macular degeneration. The New England Journal of Medicine. 2008;359(14):1456–1463. doi: 10.1056/NEJMoa0802437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Klaver CCW, Kliffen M, Van Duijn CM, et al. Genetic association of apolipoprotein E with age-related macular degeneration. American Journal of Human Genetics. 1998;63(1):200–206. doi: 10.1086/301901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Baird PN, Guida E, Chu DT, Vu HTV, Guymer RH. The ε2 and ε4 alleles of the apolipoprotein gene are associated with age-related macular degeneration. Investigative Ophthalmology and Visual Science. 2004;45(5):1311–1315. doi: 10.1167/iovs.03-1121. [DOI] [PubMed] [Google Scholar]
  • 146.Baird PN, Richardson AJ, Robman LD, et al. Apolipoprotein (APOE) gene is associated with progression of age-related macular degeneration (AMD) Human Mutation. 2006;27(4):337–342. doi: 10.1002/humu.20288. [DOI] [PubMed] [Google Scholar]
  • 147.Allikmets R, Shroyer NF, Singh N, et al. Mutation of the Stargardt disease gene (ABCR) in age-related macular degeneration. Science. 1997;277(5333):1805–1807. doi: 10.1126/science.277.5333.1805. [DOI] [PubMed] [Google Scholar]
  • 148.Allikmets R. Further evidence for an association of ABCR alleles with age-related macular degeneration. The International ABCR Screening Consortium. The American Journal of Human Genetics. 2000;67(2):487–491. doi: 10.1086/303018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Combadière C, Feumi C, Raoul W, et al. CX3CR1-dependent subretinal microglia cell accumulation is associated with cardinal features of age-related macular degeneration. Journal of Clinical Investigation. 2007;117(10):2920–2928. doi: 10.1172/JCI31692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Seddon JM, Reynolds R, Rosner B. Associations of smoking, body mass index, dietary lutein, and the LIPC genetic variant rs10468017 with advanced age-related macular degeneration. Molecular Vision. 2010;16:2412–2424. [PMC free article] [PubMed] [Google Scholar]
  • 151.Neale BM, Fagerness J, Reynolds R, et al. Genome-wide association study of advanced age-related macular degeneration identifies a role of the hepatic lipase gene (LIPC) Proceedings of the National Academy of Sciences of the United States of America. 2010;107(16):7395–7400. doi: 10.1073/pnas.0912019107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Chen W, Stambolian D, Edwards AO, et al. Genetic variants near TIMP3 and high-density lipoprotein-associated loci influence susceptibility to age-related macular degeneration. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(16):7401–7406. doi: 10.1073/pnas.0912702107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Baird PN, Chu D, Guida E, Vu HTV, Guymer R. Association of the M55L and Q192R paraoxonase gene polymorphisms with age-related macular degeneration. American Journal of Ophthalmology. 2004;138(4):665–666. doi: 10.1016/j.ajo.2004.04.053. [DOI] [PubMed] [Google Scholar]
  • 154.Tuo J, Ning B, Bojanowski CM, et al. Synergic effect of polymorphisms in ERCC6 5′ flanking region and complement factor H on age-related macular degeneration predisposition. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(24):9256–9261. doi: 10.1073/pnas.0603485103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Conley YP, Thalamuthu A, Jakobsdottir J, et al. Candidate gene analysis suggests a role for fatty acid biosynthesis and regulation of the complement system in the etiology of age-related maculopathy. Human Molecular Genetics. 2005;14(14):1991–2002. doi: 10.1093/hmg/ddi204. [DOI] [PubMed] [Google Scholar]
  • 156.Stone EM, Braun TA, Russell SR, et al. Missense variations in the fibulin 5 gene and age-related macular degeneration. The New England Journal of Medicine. 2004;351(4):346–353. doi: 10.1056/NEJMoa040833. [DOI] [PubMed] [Google Scholar]
  • 157.Schultz DW, Klein ML, Humpert AJ, et al. Analysis of the ARMD1 locus: evidence that a mutation in HEMICENTIN-1 is associated with age-related macular degeneration in a large family. Human Molecular Genetics. 2003;12(24):3315–3323. doi: 10.1093/hmg/ddg348. [DOI] [PubMed] [Google Scholar]
  • 158.Ennis S, Jomary C, Mullins R, et al. Association between the SERPING1 gene and age-related macular degeneration: a two-stage case-control study. The Lancet. 2008;372(9652):1828–1834. doi: 10.1016/S0140-6736(08)61348-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Nakata I, Yamashiro K, Yamada R, et al. Association between the serping1 gene and age-related macular degeneration and polypoidal choroidal vasculopathy in japanese. PLoS ONE. 2011;6(4) doi: 10.1371/journal.pone.0019108.e19108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Haines JL, Schnetz-Boutaud N, Schmidt S, et al. Functional candidate genes in age-related macular degeneration: significant association with VEGF, VLDLR, and LRP6. Investigative Ophthalmology and Visual Science. 2006;47(1):329–335. doi: 10.1167/iovs.05-0116. [DOI] [PubMed] [Google Scholar]
  • 161.Churchill AJ, Carter JG, Lovell HC, et al. VEGF polymorphisms are associated with neovascular age-related macular degeneration. Human Molecular Genetics. 2006;15(19):2955–2961. doi: 10.1093/hmg/ddl238. [DOI] [PubMed] [Google Scholar]
  • 162.Galan A, Ferlin A, Caretti L, et al. Association of age-related macular degeneration with polymorphisms in vascular endothelial growth factor and its receptor. Ophthalmology. 2010;117(9):1769–1774. doi: 10.1016/j.ophtha.2010.01.030. [DOI] [PubMed] [Google Scholar]
  • 163.Wang Y, Wang VM, Chan CC. The role of anti-inflammatory agents in age-related macular degeneration (AMD) treatment. Eye. 2011;25(2):127–139. doi: 10.1038/eye.2010.196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Age-Related Eye Disease Study Research Group. The Age-Related Eye Disease Study (AREDS): design implications. AREDS report no. 1. Control Clinical Trials. 1999;20(6):573–600. doi: 10.1016/s0197-2456(99)00031-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Krishnadev N, Meleth AD, Chew EY. Nutritional supplements for age-related macular degeneration. Current Opinion in Ophthalmology. 2010;21(3):184–189. doi: 10.1097/ICU.0b013e32833866ee. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Olson JH, Erie JC, Bakri SJ. Nutritional supplementation and age-related macular degeneration. Seminars in Ophthalmology. 2011;26(3):131–136. doi: 10.3109/08820538.2011.577131. [DOI] [PubMed] [Google Scholar]
  • 167.Wilkinson JT, Fraunfelder FW. Use of herbal medicines and nutritional supplements in ocular disorders: an evidence-based review. Drugs. 2011;71(18):2421–2434. doi: 10.2165/11596840-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 168.Sin HP, Liu DT, Lam DS. Lifestyle modification, nutritional and vitamins supplements for age-related macular degeneration. doi: 10.1111/j.1755-3768.2011.02357.x. Acta Ophthalmologica. In press. [DOI] [PubMed] [Google Scholar]
  • 169.Zeng S, Hernández J, Mullins RF. Effects of antioxidant components of AREDS vitamins and zinc ions on endothelial cell activation: implications for macular degeneration. Investigative Ophthalmology & Visual Science. 2012;53(2):1041–1047. doi: 10.1167/iovs.11-8531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Weigert G, Kaya S, Pemp B, et al. Effects of lutein supplementation on macular pigment optical density and visual acuity in patients with age-related macular degeneration. Investigative Ophthalmology & Visual Science. 2011;52(11):8174–8178. doi: 10.1167/iovs.11-7522. [DOI] [PubMed] [Google Scholar]
  • 171.Kijlstra A, Tian Y, Kelly ER, Berendschot TT. Lutein: more than just a filter for blue light. Progress in Retinal and Eye Research. 2012;31(4):303–315. doi: 10.1016/j.preteyeres.2012.03.002. [DOI] [PubMed] [Google Scholar]
  • 172.Penfold PL, Wong JG, Gyory J, Billson FA. Effects of triamcinolone acetonide on microglial morphology and quantitative expression of MHC-II in exudative age-related macular degeneration. Clinical and Experimental Ophthalmology. 2001;29(3):188–192. doi: 10.1046/j.1442-9071.2001.00407.x. [DOI] [PubMed] [Google Scholar]
  • 173.Becerra EM, Morescalchi F, Gandolfo F, et al. Clinical evidence of intravitreal triamcinolone acetonide in the management of age-related macular degeneration. Current drug targets. 2011;12(2):149–172. doi: 10.2174/138945011794182746. [DOI] [PubMed] [Google Scholar]
  • 174.Nussenblatt RB, Byrnes G, Sen HN, et al. A randomized pilot study of systemic immunosuppression in the treatment of age-related macular degeneration with choroidal neovascularization. Retina. 2010;30(10):1579–1587. doi: 10.1097/IAE.0b013e3181e7978e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Arias L, Caminal JM, Badia MB, Rubio MJ, Catala J, Pujol O. Intravitreal infliximab in patients with macular degeneration who are nonresponders to antivascular endothelial growth factor therapy. Retina. 2010;30(10):1601–1608. doi: 10.1097/IAE.0b013e3181e9f942. [DOI] [PubMed] [Google Scholar]
  • 176.Chakravarthy U, Soubrane G, Bandello F, et al. Evolving European guidance on the medical management of neovascular age related macular degeneration. British Journal of Ophthalmology. 2006;90(9):1188–1196. doi: 10.1136/bjo.2005.082255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Wormald R, Evans J, Smeeth L, Henshaw K. Photodynamic therapy for neovascular age-related macular degeneration. Cochrane Database of Systematic Reviews. 2007;(3):p. CD002030. doi: 10.1002/14651858.CD002030.pub3. [DOI] [PubMed] [Google Scholar]
  • 178.Vedula SS, Krzystolik MG. Antiangiogenic therapy with anti-vascular endothelial growth factor modalities for neovascular age-related macular degeneration. Cochrane Database of Systematic Reviews. 2008;(2):p. CD005139. doi: 10.1002/14651858.CD005139.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Colquitt JL, Jones J, Tan SC, Takeda A, Clegg AJ, Price A. Ranibizumab and pegaptanib for the treatment of age-related macular degeneration: a systematic review and economic evaluation. Health Technology Assessment. 2008;12(16) doi: 10.3310/hta12160. [DOI] [PubMed] [Google Scholar]
  • 180.Mitchell P, Korobelnik JF, Lanzetta P, et al. Ranibizumab (Lucentis) in neovascular age-related macular degeneration: evidence from clinical trials. British Journal of Ophthalmology. 2010;94(1):2–13. doi: 10.1136/bjo.2009.159160. [DOI] [PubMed] [Google Scholar]
  • 181.Martin DF, Maguire MG, Ying GS, Grunwald JE, Fine SL, Jaffe GJ. Ranibizumab and bevacizumab for neovascular age-related macular degeneration. The New England Journal of Medicine. 2011;364(20):1897–1908. doi: 10.1056/NEJMoa1102673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Rosenfeld PJ. Bevacizumab versus ranibizumab for AMD. The New England Journal of Medicine. 2011;364(20):1966–1967. doi: 10.1056/NEJMe1103334. [DOI] [PubMed] [Google Scholar]
  • 183.Ziemssen F, Sobolewska B. Therapeutic efficacy of bevacizumab for age-related macular degeneration: what are the implications of CATT for routine management? Drugs & Aging. 2011;28(11):853–865. doi: 10.2165/11594720-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 184.Davis J, Olsen TW, Stewart M, Sternberg Jr P. How the comparison of age-related macular degeneration treatments trial results will impact clinical care. American Journal of Ophthalmology. 2011;152(4):509–514. doi: 10.1016/j.ajo.2011.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Csaky KG. Comparison of age-related macular degeneration treatment trials: what did we learn? Retina. 2012;32(3):413–416. doi: 10.1097/IAE.0B013E3182493C05. [DOI] [PubMed] [Google Scholar]
  • 186.Martin DF, Maguire MG, Fine SL, et al. Ranibizumab and bevacizumab for treatment of neovascular age-related macular degeneration: two-year results. Ophthalmology. 2012;119(7):1388–1398. doi: 10.1016/j.ophtha.2012.03.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Clinical Trials.gov identifier: NCT00766649, “Sirolimus to treat geographic atrophy associated with age-related macular degeneration”, 2010, http://www.clinicaltrial.gov/ct2/show/NCT00766649.
  • 188.Patel M, Chan CC. Immunopathological aspects of age-related macular degeneration. Seminars in Immunopathology. 2008;30(2):97–110. doi: 10.1007/s00281-008-0112-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Augustin AJ, Puls S, Offermann I. Triple therapy for choroidal neovascularization due to age-related macular degeneration: verteporfin PDT, bevacizumab, and dexamethasone. Retina. 2007;27(2):133–140. doi: 10.1097/IAE.0b013e3180323de7. [DOI] [PubMed] [Google Scholar]
  • 190.Bakri SJ, Couch SM, McCannel CA, Edwards AO. Same-day triple therapy with photodynamic therapy, intravitreal dexamethasone, and bevacizumab in wet age-related macular degeneration. Retina. 2009;29(5):573–578. doi: 10.1097/IAE.0b013e3181a46a8a. [DOI] [PubMed] [Google Scholar]
  • 191.Ehmann D, García R. Triple therapy for neovascular age-related macular degeneration (verteporfin photodynamic therapy, intravitreal dexamethasone, and intravitreal bevacizumab) Canadian Journal of Ophthalmology. 2010;45(1):36–40. doi: 10.3129/i09-243. [DOI] [PubMed] [Google Scholar]
  • 192.Forte R, Bonavolontà P, Benayoun Y, Adenis JP, Robert PY. Intravitreal ranibizumab and bevacizumab in combination with full-fluence verteporfin therapy and dexamethasone for exudative age-related macular degeneration. Ophthalmic Research. 2011;45(3):129–134. doi: 10.1159/000318877. [DOI] [PubMed] [Google Scholar]
  • 193.Sivaprasad S, Patra S, Dacosta J, et al. A pilot study on the combination treatment of reduced-fluence photodynamic therapy, intravitreal ranibizumab, intravitreal dexamethasone and oral minocycline for neovascular age-related macular degeneration. Ophthalmologica. 2011;225(4):200–206. doi: 10.1159/000322363. [DOI] [PubMed] [Google Scholar]
  • 194.Singer M, Patel SS, Harvey SU, Liu CC, Li XY, Whitcup SM. Dexamethasone intravitreal implant (DEX implant) alone and as an adjunct to ranibizumab for the treatment of choroidal neovascularization (CNV) secondary to age-related macular degeneration (AMD). Proceedings of the Association for Research in Vision and Ophthalmology, Poster Presentation 3536/A334; 2011. [Google Scholar]
  • 195.Schmidt-Erfurth U, Michels S, Barbazetto I, Laqua H. Photodynamic effects on choroidal neovascularization and physiological choroid. Investigative Ophthalmology and Visual Science. 2002;43(3):830–841. [PubMed] [Google Scholar]
  • 196.Michels S, Schmidt-Erfurth U. Sequence of early vascular events after photodynamic therapy. Investigative Ophthalmology and Visual Science. 2003;44(5):2147–2154. doi: 10.1167/iovs.02-0604. [DOI] [PubMed] [Google Scholar]
  • 197.Schmidt-Erfurth U, Niemeyer M, Geitzenauer W, Michels S. Time course and morphology of vascular effects associated with photodynamic therapy. Ophthalmology. 2005;112(12):2061–2069. doi: 10.1016/j.ophtha.2005.09.007. [DOI] [PubMed] [Google Scholar]
  • 198.Brantley MA, Jr., Fang AM, King JM, Tewari A, Kymes SM, Shiels A. Association of complement factor H and LOC387715 genotypes with response of exudative age-related macular degeneration to intravitreal bevacizumab. Ophthalmology. 2007;114(12):2168–2173. doi: 10.1016/j.ophtha.2007.09.008. [DOI] [PubMed] [Google Scholar]
  • 199.Seitsonen SP, Järvelä IE, Meri S, Tommila PV, Ranta PH, Immonen IJ. The effect of complement factor H Y402H polymorphism on the outcome of photodynamic therapy in age-related macular degeneration. European Journal of Ophthalmology. 2007;17(6):943–949. doi: 10.1177/112067210701700612. [DOI] [PubMed] [Google Scholar]
  • 200.Parmeggiani F, Costagliola C, Gemmati D, et al. Predictive role of coagulation-balance gene polymorphisms in the efficacy of photodynamic therapy with verteporfin for classic choroidal neovascularization secondary to age-related macular degeneration. Pharmacogenetics and Genomics. 2007;17(12):1039–1046. doi: 10.1097/FPC.0b013e3282f12a4e. [DOI] [PubMed] [Google Scholar]
  • 201.Ranchod TM, Guercio JR, Ying GS, Brucker AJ, Stoltz RA. Effect of aspirin therapy on photodynamic therapy with verteporfin for choroidal neovascularization. Retina. 2008;28(5):711–716. doi: 10.1097/IAE.0b013e31816079c3. [DOI] [PubMed] [Google Scholar]
  • 202.Parmeggiani F, Costagliola C, Gemmati D, et al. Coagulation gene predictors of photodynamic therapy for occult choroidal neovascularization in age-related macular degeneration. Investigative Ophthalmology and Visual Science. 2008;49(7):3100–3106. doi: 10.1167/iovs.07-1654. [DOI] [PubMed] [Google Scholar]
  • 203.Goverdhan SV, Hannan S, Newsom RB, Luff AJ, Griffiths H, Lotery AJ. An analysis of the CFH Y402H genotype in AMD patients and controls from the UK, and response to PDT treatment. Eye. 2008;22(6):849–854. doi: 10.1038/sj.eye.6702830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Chowers I, Meir T, Lederman M, et al. Sequence variants in HTRA1 and LOC387715/ARMS2 and phenotype and response to photodynamic therapy in neovascular age-related macular degeneration in populations from Israel. Molecular Vision. 2008;14:2263–2271. [PMC free article] [PubMed] [Google Scholar]
  • 205.Lee AY, Raya AK, Kymes SM, Shiels A, Brantley MA., Jr. Pharmacogenetics of complement factor H (Y402H) and treatment of exudative age-related macular degeneration with ranibizumab. British Journal of Ophthalmology. 2009;93(5):610–613. doi: 10.1136/bjo.2008.150995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Brantley MA, Jr., Edelstein SL, King JM, et al. Association of complement factor H and LOC387715 genotypes with response of exudative age-related macular degeneration to photodynamic therapy. Eye. 2009;23(3):626–631. doi: 10.1038/eye.2008.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Feng X, Xiao J, Longville B, et al. Complement factor H Y402H and C-reactive protein polymorphism and photodynamic therapy response in age-related macular degeneration. Ophthalmology. 2009;116(10):1908–e1. doi: 10.1016/j.ophtha.2009.03.011. [DOI] [PubMed] [Google Scholar]
  • 208.Parmeggiani F, Gemmati D, Costagliola C, Sebastiani A, Incorvaia C. Predictive role of C677T MTHFR polymorphism in variable efficacy of photodynamic therapy for neovascular age-related macular degeneration. Pharmacogenomics. 2009;10(1):81–95. doi: 10.2217/14622416.10.1.81. [DOI] [PubMed] [Google Scholar]
  • 209.Parmeggiani F, Gemmati D, Costagliola C, Sebastiani A, Incorvaia C. Predictive role of gene polymorphisms affecting thrombin-generation pathway in variable efficacy of photodynamic therapy for neovascular age-related macular degeneration. Recent Patents on DNA and Gene Sequences. 2009;3(2):114–122. doi: 10.2174/187221509788654151. [DOI] [PubMed] [Google Scholar]
  • 210.Teper SJ, Nowinska A, Pilat J, Palucha A, Wylegala E. Involvement of genetic factors in the response to a variable-dosing ranibizumab treatment regimen for age-related macular degeneration. Molecular Vision. 2010;16:2598–2604. [PMC free article] [PubMed] [Google Scholar]
  • 211.Imai D, Mori K, Horie-Inoue K, et al. CFH, VEGF, and PEDF genotypes and the response to intravitreous injection of bevacizumab for the treatment of age-related macular degeneration. Journal of Ocular Biology, Diseases, and Informatics. 2010;3(2):53–59. doi: 10.1007/s12177-010-9055-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Immonen I, Seitsonen S, Tommila P, et al. Vascular endothelial growth factor gene variation and the response to photodynamic therapy in age-related macular degeneration. Ophthalmology. 2010;117(1):103–108. doi: 10.1016/j.ophtha.2009.06.037. [DOI] [PubMed] [Google Scholar]
  • 213.Shastry BS. Genetic diversity and medicinal drug response in eye care. Graefe’s Archive for Clinical and Experimental Ophthalmology. 2010;248(8):1057–1061. doi: 10.1007/s00417-010-1333-x. [DOI] [PubMed] [Google Scholar]
  • 214.Kloeckener-Gruissem B, Barthelmes D, Labs S, et al. Genetic association with response to intravitreal ranibizumab in patients with neovascular AMD. Investigative Ophthalmology & Visual Science. 2011;52(7):4694–4702. doi: 10.1167/iovs.10-6080. [DOI] [PubMed] [Google Scholar]
  • 215.Nischler C, Oberkofler H, Ortner C, et al. Complement factor H Y402H gene polymorphism and response to intravitreal bevacizumab in exudative age-related macular degeneration. Acta Ophthalmologica. 2011;89(4):e344–e349. doi: 10.1111/j.1755-3768.2010.02080.x. [DOI] [PubMed] [Google Scholar]
  • 216.Parmeggiani F, Costagliola C, Incorvaia C, Sebastiani A, Gemmati D. Pharmacogenetic aspects in therapeutic management of subfoveal choroidal neovascularisation: role of factor XIII-A 185 T-allele. Current drug targets. 2011;12(2):138–148. doi: 10.2174/138945011794182773. [DOI] [PubMed] [Google Scholar]
  • 217.Tsuchihashi T, Mori K, Horie-Inoue K, et al. Complement factor H and high-temperature requirement A-1 genotypes and treatment response of age-related macular degeneration. Ophthalmology. 2011;118(1):93–100. doi: 10.1016/j.ophtha.2010.04.007. [DOI] [PubMed] [Google Scholar]
  • 218.Ramo K, Cashman SM, Kumar-Singh R. Evaluation of adenovirus-delivered human CD59 as a potential therapy for AMD in a model of human membrane attack complex formation on murine RPE. Investigative Ophthalmology and Visual Science. 2008;49(9):4126–4136. doi: 10.1167/iovs.08-2025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Gandhi J, Cashman SM, Kumar-Singh R. Soluble CD59 expressed from an adenovirus in vivo is a potent inhibitor of complement deposition on murine liver vascular endothelium. PLoS ONE. 2011;6(6) doi: 10.1371/journal.pone.0021621.e21621 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Sweigard JH, Cashman SM, Kumar-Singh R. Adenovirus-mediated delivery of CD46 attenuates the alternative complement pathway on RPE: implications for age-related macular degeneration. Gene Therapy. 2011;18(6):613–621. doi: 10.1038/gt.2011.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Kuno N, Fujii S. Biodegradable intraocular therapies for retinal disorders: progress to date. Drugs and Aging. 2010;27(2):117–134. doi: 10.2165/11530970-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 222.London NJS, Chiang A, Haller JA. The dexamethasone drug delivery system: indications and evidence. Advances in Therapy. 2011;28(5):351–366. doi: 10.1007/s12325-011-0019-z. [DOI] [PubMed] [Google Scholar]
  • 223.de Oliveira Dias JR, Rodrigues EB, Maia M, Magalhães O, Penha FM, Farah ME. Cytokines in neovascular age-related macular degeneration: fundamentals of targeted combination therapy. British Journal of Ophthalmology. 2011;95(12):1631–1637. doi: 10.1136/bjo.2010.186361. [DOI] [PubMed] [Google Scholar]
  • 224.Packard BD, Weiler JM. Steroids inhibit activation of the alternative-amplification pathway of complement. Infection and Immunity. 1983;40(3):1011–1014. doi: 10.1128/iai.40.3.1011-1014.1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Barnes PJ. Anti-inflammatory actions of glucocorticoids: molecular mechanisms. Clinical Science. 1998;94(6):557–572. doi: 10.1042/cs0940557. [DOI] [PubMed] [Google Scholar]

Articles from Mediators of Inflammation are provided here courtesy of Wiley

RESOURCES